Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 120(43): e2307203120, 2023 Oct 24.
Artigo em Inglês | MEDLINE | ID: mdl-37844219

RESUMO

The TGF-beta signals Vg1 (Dvr1/Gdf3) and Nodal form heterodimers to induce vertebrate mesendoderm. The Vg1 proprotein is a monomer retained in the endoplasmic reticulum (ER) and is processed and secreted upon heterodimerization with Nodal, but the mechanisms underlying Vg1 biogenesis are largely elusive. Here, we clarify the mechanisms underlying Vg1 retention, processing, secretion, and signaling and introduce a Synthetic Processing (SynPro) system that enables the programmed cleavage of ER-resident and extracellular proteins. First, we find that Vg1 can be processed by intra- or extracellular proteases. Second, Vg1 can be processed without Nodal but requires Nodal for secretion and signaling. Third, Vg1-Nodal signaling activity requires Vg1 processing, whereas Nodal can remain unprocessed. Fourth, Vg1 employs exposed cysteines, glycosylated asparagines, and BiP chaperone-binding motifs for monomer retention in the ER. These observations suggest two mechanisms for rapid mesendoderm induction: Chaperone-binding motifs help store Vg1 as an inactive but ready-to-heterodimerize monomer in the ER, and the flexibility of Vg1 processing location allows efficient generation of active heterodimers both intra- and extracellularly. These results establish SynPro as an in vivo processing system and define molecular mechanisms and motifs that facilitate the generation of active TGF-beta heterodimers.


Assuntos
Padronização Corporal , Fator de Crescimento Transformador beta , Animais , Fator de Crescimento Transformador beta/metabolismo , Vertebrados/metabolismo , Transdução de Sinais
2.
Methods ; 157: 3-14, 2019 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-30593865

RESUMO

Tissues such as brain, muscle, and bone differ greatly not only in their biological functions but also in their mechanical properties. Brain is far softer than muscle while bone is the stiffest tissue. Stiffness of extracellular microenvironments affects fundamental cell biological processes such as polarization and DNA replication, which affect nuclear size, shape, and levels of nuclear proteins such as the lamins that modulate gene expression. Reductionist approaches have helped dissect the effects of matrix mechanics away from confounding biochemical signals. Here, we summarize materials and methods for synthesizing and characterizing soft and stiff synthetic hydrogels widely used for mechanobiological studies. Such gels are also easily made to mimic the mechanical heterogeneity of fibrotic tissues. We further describe a nano-thin collagen fiber system, which enables control of anisotropy in addition to stiffness. With the different systems, we illustrate the effects of matrix mechanics on nuclear size, shape, and proteins including the lamins.


Assuntos
Biologia Celular , Técnicas Citológicas/métodos , Matriz Extracelular/ultraestrutura , Anisotropia , Matriz Extracelular/genética , Regulação da Expressão Gênica/genética , Hidrogéis/química , Fenômenos Mecânicos
3.
Nat Commun ; 8(1): 2212, 2017 12 20.
Artigo em Inglês | MEDLINE | ID: mdl-29263378

RESUMO

G-protein-coupled receptors (GPCRs) are the largest and most diverse group of membrane receptors in eukaryotes and detect a wide array of cues in the human body. Here we describe a molecular device that couples CRISPR-dCas9 genome regulation to diverse natural and synthetic extracellular signals via GPCRs. We generate alternative architectures for fusing CRISPR to GPCRs utilizing the previously reported design, Tango, and our design, ChaCha. Mathematical modeling suggests that for the CRISPR ChaCha design, multiple dCas9 molecules can be released across the lifetime of a GPCR. The CRISPR ChaCha is dose-dependent, reversible, and can activate multiple endogenous genes simultaneously in response to extracellular ligands. We adopt the design to diverse GPCRs that sense a broad spectrum of ligands, including synthetic compounds, chemokines, mitogens, fatty acids, and hormones. This toolkit of CRISPR-coupled GPCRs provides a modular platform for rewiring diverse ligand sensing to targeted genome regulation for engineering cellular functions.


Assuntos
Sistemas CRISPR-Cas , Engenharia Celular/métodos , Receptores Acoplados a Proteínas G , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas , Células HEK293 , Humanos , Ligantes , Modelos Teóricos
4.
Nat Mater ; 14(9): 951-60, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26168347

RESUMO

Scarring is a long-lasting problem in higher animals, and reductionist approaches could aid in developing treatments. Here, we show that copolymerization of collagen I with polyacrylamide produces minimal matrix models of scars (MMMS), in which fractal-fibre bundles segregate heterogeneously to the hydrogel subsurface. Matrix stiffens locally-as in scars-while allowing separate control over adhesive-ligand density. The MMMS elicits scar-like phenotypes from mesenchymal stem cells (MSCs): cells spread and polarize quickly, increasing nucleoskeletal lamin-A yet expressing the 'scar marker' smooth muscle actin (SMA) more slowly. Surprisingly, expression responses to MMMS exhibit less cell-to-cell noise than homogeneously stiff gels. Such differences from bulk-average responses arise because a strong SMA repressor, NKX2.5, slowly exits the nucleus on rigid matrices. NKX2.5 overexpression overrides rigid phenotypes, inhibiting SMA and cell spreading, whereas cytoplasm-localized NKX2.5 mutants degrade in well-spread cells. MSCs thus form a 'mechanical memory' of rigidity by progressively suppressing NKX2.5, thereby elevating SMA in a scar-like state.


Assuntos
Núcleo Celular/metabolismo , Cicatriz/metabolismo , Matriz Extracelular/química , Proteínas de Homeodomínio/metabolismo , Células-Tronco Mesenquimais/metabolismo , Nicho de Células-Tronco , Fatores de Transcrição/metabolismo , Resinas Acrílicas/química , Actinas/metabolismo , Transporte Ativo do Núcleo Celular , Animais , Núcleo Celular/patologia , Cicatriz/patologia , Colágeno Tipo I/química , Proteína Homeobox Nkx-2.5 , Camundongos , Modelos Biológicos
6.
Biophys J ; 107(11): 2734-43, 2014 Dec 02.
Artigo em Inglês | MEDLINE | ID: mdl-25468352

RESUMO

Mechanotransduction pathways convert forces that stress and strain structures within cells into gene expression levels that impact development, homeostasis, and disease. The levels of some key structural proteins in the nucleus, cytoskeleton, or extracellular matrix have been recently reported to scale with tissue- and cell-level forces or mechanical properties such as stiffness, and so the mathematics of mechanotransduction becomes important to understand. Here, we show that if a given structural protein positively regulates its own gene expression, then stresses need only inhibit degradation of that protein to achieve stable, mechanosensitive gene expression. This basic use-it-or-lose-it module is illustrated by application to meshworks of nuclear lamin A, minifilaments of myosin II, and extracellular matrix collagen fibers­all of which possess filamentous coiled-coil/supercoiled structures. Past experiments not only suggest that tension suppresses protein degradation mediated and/or initiated by various enzymes but also that transcript levels vary with protein levels because key transcription factors are regulated by these structural proteins. Coupling between modules occurs within single cells and between cells in tissue, as illustrated during embryonic heart development where cardiac fibroblasts make collagen that cardiomyocytes contract. With few additional assumptions, the basic module has sufficient physics to control key structural genes in both development and disease.


Assuntos
Redes Reguladoras de Genes , Mecanotransdução Celular , Proteínas/metabolismo , Estresse Mecânico , Biologia de Sistemas , Animais , Colágeno/metabolismo , Matriz Extracelular/metabolismo , Retroalimentação Fisiológica , Fibroblastos/metabolismo , Lamina Tipo A/metabolismo , Camundongos , Miócitos Cardíacos/metabolismo , Miosinas/metabolismo , Células NIH 3T3
7.
Curr Biol ; 24(16): 1909-17, 2014 Aug 18.
Artigo em Inglês | MEDLINE | ID: mdl-25127216

RESUMO

Tissue microenvironments are characterized not only in terms of chemical composition but also by collective properties such as stiffness, which influences the contractility of a cell, its adherent morphology, and even differentiation. The nucleoskeletal protein lamin-A,C increases with matrix stiffness, confers nuclear mechanical properties, and influences differentiation of mesenchymal stem cells (MSCs), whereas B-type lamins remain relatively constant. Here we show in single-cell analyses that matrix stiffness couples to myosin-II activity to promote lamin-A,C dephosphorylation at Ser22, which regulates turnover, lamina physical properties, and actomyosin expression. Lamin-A,C phosphorylation is low in interphase versus dividing cells, and its levels rise with states of nuclear rounding in which myosin-II generates little to no tension. Phosphorylated lamin-A,C localizes to nucleoplasm, and phosphorylation is enriched on lamin-A,C fragments and is suppressed by a cyclin-dependent kinase (CDK) inhibitor. Lamin-A,C knockdown in primary MSCs suppresses transcripts predominantly among actomyosin genes, especially in the serum response factor (SRF) pathway. Levels of myosin-IIA thus parallel levels of lamin-A,C, with phosphosite mutants revealing a key role for phosphoregulation. In modeling the system as a parsimonious gene circuit, we show that tension-dependent stabilization of lamin-A,C and myosin-IIA can suitably couple nuclear and cell morphology downstream of matrix mechanics.


Assuntos
Matriz Extracelular/metabolismo , Lamina Tipo A/genética , Células-Tronco Mesenquimais/metabolismo , Miosina não Muscular Tipo IIA/genética , Diferenciação Celular , Elasticidade , Retroalimentação Fisiológica , Humanos , Lamina Tipo A/metabolismo , Miosina não Muscular Tipo IIA/metabolismo , Fosforilação , Análise de Célula Única
8.
Curr Biol ; 24(10): R495-501, 2014 May 19.
Artigo em Inglês | MEDLINE | ID: mdl-24845682

RESUMO

Early in embryogenesis, the heart begins its rhythmic contractions as a tube that helps perfuse the nascent vasculature, but the embryonic heart soon changes shape and mechanical properties, like many other developing organs. A key question in the field is whether stresses in development impact the underlying gene circuits and, if so, how? Here, we attempt to address this question as we review the mechanical maturation of heart - and, to a limited extent, lung and blood - with a focus on a few key abundant structural proteins whose expression dynamics have been suggested to be directly sensitive to mechanical stress. In heart maturation, proliferating fibroblasts deposit increasing amounts of collagenous matrix in parallel with cardiomyocytes expressing more sarcomeric proteins that increase the contractile stress and strength of the tissue, which in turn pumps more blood at higher stress throughout the developing vasculature. Feedback of beating cardiomyocytes on the expression of matrix by fibroblasts seems a reasonable model, with both synthesis and turnover of matrix and contractile elements achieving a suitable balance. Based on emerging evidence for coiled-coil biopolymers that are tension-stabilized against degradation, a minimal network model of a dynamic cell-matrix interaction is proposed. This same concept is extended to nuclear mechanics as regulated by stress on the nuclear structural proteins called lamins, which are examined in part because of the prominence of mutations in these coiled-coil proteins in diseases of the heart, amongst other organs/tissues. Variations in lamin levels during development and across adult tissues are to some extent known and appear to correlate with extracellular matrix mechanics, which we illustrate across heart, lung, and blood development. The formal perspective here on the mechanochemistry of tissue development and homeostasis could provide a useful framework for 'big data' quantitative biology, particularly of stress-sensitive differentiation, maturation, and disease processes.


Assuntos
Coração/embriologia , Mecanotransdução Celular , Estresse Mecânico , Animais , Sangue/metabolismo , Matriz Extracelular/metabolismo , Fibroblastos/metabolismo , Humanos , Laminas/metabolismo , Pulmão/embriologia , Miócitos Cardíacos/metabolismo
9.
Curr Opin Biotechnol ; 28: 46-50, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24832074

RESUMO

Recent experiments have revealed that stem cells respond to biophysical cues as well as numerous biochemical factors. Nanoscale properties at the cell-matrix interface that appear to affect adherent stem cells range from matrix elasticity to porosity-dependent matrix tethering and geometry of adhesive linkages. Some stem cells can also remodel their immediate environment to influence phenotype, but this depends on matrix-material properties such as covalent bonding and soft versus hard materials. Efforts to combine both matrix instructions and active cell feedback are required to properly direct stem cell behavior. Comparisons to tissues will be increasingly key and have begun to reveal remodeling of nuclear factors that influence epigenetics.


Assuntos
Diferenciação Celular , Nanoestruturas/química , Células-Tronco/citologia , Animais , Adesão Celular , Elasticidade , Hidrogel de Polietilenoglicol-Dimetacrilato/química , Oligopeptídeos/química , Oligopeptídeos/metabolismo , Porosidade
11.
J Cell Biol ; 204(5): 669-82, 2014 Mar 03.
Artigo em Inglês | MEDLINE | ID: mdl-24567359

RESUMO

Cell migration through solid tissue often involves large contortions of the nucleus, but biological significance is largely unclear. The nucleoskeletal protein lamin-A varies both within and between cell types and was shown here to contribute to cell sorting and survival in migration through constraining micropores. Lamin-A proved rate-limiting in 3D migration of diverse human cells that ranged from glioma and adenocarcinoma lines to primary mesenchymal stem cells (MSCs). Stoichiometry of A- to B-type lamins established an activation barrier, with high lamin-A:B producing extruded nuclear shapes after migration. Because the juxtaposed A and B polymer assemblies respectively conferred viscous and elastic stiffness to the nucleus, subpopulations with different A:B levels sorted in 3D migration. However, net migration was also biphasic in lamin-A, as wild-type lamin-A levels protected against stress-induced death, whereas deep knockdown caused broad defects in stress resistance. In vivo xenografts proved consistent with A:B-based cell sorting, and intermediate A:B-enhanced tumor growth. Lamins thus impede 3D migration but also promote survival against migration-induced stresses.


Assuntos
Movimento Celular/fisiologia , Lamina Tipo A/fisiologia , Lamina Tipo B/fisiologia , Apoptose , Linhagem Celular Tumoral , Núcleo Celular/ultraestrutura , Forma do Núcleo Celular , Sobrevivência Celular , Técnicas de Silenciamento de Genes , Humanos , Lamina Tipo A/química , Lamina Tipo A/genética , Lamina Tipo B/química , Lamina Tipo B/genética , Estrutura Terciária de Proteína
12.
Cell Stem Cell ; 14(1): 81-93, 2014 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-24268694

RESUMO

Self-renewal and differentiation of stem cells depend on asymmetric division and polarized motility processes that in other cell types are modulated by nonmuscle myosin-II (MII) forces and matrix mechanics. Here, mass spectrometry-calibrated intracellular flow cytometry of human hematopoiesis reveals MIIB to be a major isoform that is strongly polarized in hematopoietic stem cells and progenitors (HSC/Ps) and thereby downregulated in differentiated cells via asymmetric division. MIIA is constitutive and activated by dephosphorylation during cytokine-triggered differentiation of cells grown on stiff, endosteum-like matrix, but not soft, marrow-like matrix. In vivo, MIIB is required for generation of blood, while MIIA is required for sustained HSC/P engraftment. Reversible inhibition of both isoforms in culture with blebbistatin enriches for long-term hematopoietic multilineage reconstituting cells by 5-fold or more as assessed in vivo. Megakaryocytes also become more polyploid, producing 4-fold more platelets. MII is thus a multifunctional node in polarized division and niche sensing.


Assuntos
Diferenciação Celular , Movimento Celular , Hematopoese/fisiologia , Células-Tronco Hematopoéticas/citologia , Contração Muscular/fisiologia , Miosina não Muscular Tipo IIA/metabolismo , Miosina não Muscular Tipo IIB/metabolismo , Apoptose , Western Blotting , Técnicas de Cultura de Células , Linhagem da Célula , Proliferação de Células , Citometria de Fluxo , Células-Tronco Hematopoéticas/fisiologia , Compostos Heterocíclicos de 4 ou mais Anéis/farmacologia , Humanos , Fosforilação , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Nicho de Células-Tronco/fisiologia
13.
Science ; 341(6149): 1240104, 2013 Aug 30.
Artigo em Inglês | MEDLINE | ID: mdl-23990565

RESUMO

Tissues can be soft like fat, which bears little stress, or stiff like bone, which sustains high stress, but whether there is a systematic relationship between tissue mechanics and differentiation is unknown. Here, proteomics analyses revealed that levels of the nucleoskeletal protein lamin-A scaled with tissue elasticity, E, as did levels of collagens in the extracellular matrix that determine E. Stem cell differentiation into fat on soft matrix was enhanced by low lamin-A levels, whereas differentiation into bone on stiff matrix was enhanced by high lamin-A levels. Matrix stiffness directly influenced lamin-A protein levels, and, although lamin-A transcription was regulated by the vitamin A/retinoic acid (RA) pathway with broad roles in development, nuclear entry of RA receptors was modulated by lamin-A protein. Tissue stiffness and stress thus increase lamin-A levels, which stabilize the nucleus while also contributing to lineage determination.


Assuntos
Diferenciação Celular , Elasticidade , Lamina Tipo A/metabolismo , Células-Tronco Mesenquimais/citologia , Osteogênese , Estresse Mecânico , Adipogenia , Animais , Colágeno/análise , Colágeno/química , Colágeno/metabolismo , Matriz Extracelular/química , Matriz Extracelular/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Lamina Tipo A/química , Lamina Tipo A/genética , Camundongos , Modelos Biológicos , Lâmina Nuclear/metabolismo , Osteogênese/genética , Conformação Proteica , Proteoma , Transcrição Gênica , Tretinoína/metabolismo , Vitamina A/metabolismo
14.
J Cell Biol ; 199(4): 669-83, 2012 Nov 12.
Artigo em Inglês | MEDLINE | ID: mdl-23128239

RESUMO

On rigid surfaces, the cytoskeleton of migrating cells is polarized, but tissue matrix is normally soft. We show that nonmuscle MIIB (myosin-IIB) is unpolarized in cells on soft matrix in 2D and also within soft 3D collagen, with rearward polarization of MIIB emerging only as cells migrate from soft to stiff matrix. Durotaxis is the tendency of cells to crawl from soft to stiff matrix, and durotaxis of primary mesenchymal stem cells (MSCs) proved more sensitive to MIIB than to the more abundant and persistently unpolarized nonmuscle MIIA (myosin-IIA). However, MIIA has a key upstream role: in cells on soft matrix, MIIA appeared diffuse and mobile, whereas on stiff matrix, MIIA was strongly assembled in oriented stress fibers that MIIB then polarized. The difference was caused in part by elevated phospho-S1943-MIIA in MSCs on soft matrix, with site-specific mutants revealing the importance of phosphomoderated assembly of MIIA. Polarization is thus shown to be a highly regulated compass for mechanosensitive migration.


Assuntos
Citoesqueleto/metabolismo , Matriz Extracelular/metabolismo , Miosina não Muscular Tipo IIA/metabolismo , Miosina não Muscular Tipo IIB/metabolismo , Movimento Celular , Polaridade Celular , Células Cultivadas , Humanos , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Fosforilação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...